EP3209296B1 - Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors - Google Patents

Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors Download PDF

Info

Publication number
EP3209296B1
EP3209296B1 EP15853280.4A EP15853280A EP3209296B1 EP 3209296 B1 EP3209296 B1 EP 3209296B1 EP 15853280 A EP15853280 A EP 15853280A EP 3209296 B1 EP3209296 B1 EP 3209296B1
Authority
EP
European Patent Office
Prior art keywords
tert
oxo
butyl
pyridin
carboxylate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP15853280.4A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP3209296A4 (en
EP3209296A1 (en
Inventor
Mark E. Scott
David Guerin
Danielle MOLINARI
Sam Kattar
Peter Fuller
Christopher Dinsmore
Norman Kong
Yunfeng BAI
Jiamin FU
Yumei Liu
Zhixiang Zheng
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Sharp and Dohme LLC
Original Assignee
Merck Sharp and Dohme LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Sharp and Dohme LLC filed Critical Merck Sharp and Dohme LLC
Publication of EP3209296A1 publication Critical patent/EP3209296A1/en
Publication of EP3209296A4 publication Critical patent/EP3209296A4/en
Application granted granted Critical
Publication of EP3209296B1 publication Critical patent/EP3209296B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/5381,4-Oxazines, e.g. morpholine ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/554Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one sulfur as ring hetero atoms, e.g. clothiapine, diltiazem
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Protein kinases are a group of enzymes that regulate the activity of their target proteins by the addition of phosphate groups to the protein substrate. Kinases play an essential role in many physiological processes including cell division, differentiation, cellular homeostasis and signal transduction. Kinases can be subdivided by their target into Serine/Threonine kinases and Tyrosine kinases. Tyrosine kinases are further subdivided into receptor tyrosine kinases and non-receptor tyrosine kinases. The mammalian Janus kinase (JAK) family members are non-receptor tyrosine kinases.
  • JAK Japanese Janus kinase
  • JAK1 JAK1, JAK2, JAK3 and TYK2.
  • JAK1, JAK2 and TYK2 are universally expressed, whereas JAK3 expression is limited to hematopoetic cells.
  • the JAK family is involved in intracellular signal transduction from >70 different cytokines. Cytokines bind to their cell surface receptors resulting in receptor dimerization and subsequent activation/phosphorylation of JAK tyrosine kinases.
  • the JAKs are either constitutively associated with the receptor or are recruited upon cytokine binding. Specific tyrosine residues on the receptor are then phosphorylated by activated JAKs and serve as docking sites for STAT proteins. STATs are phosphorylated by JAKs, dimerize, then translocate to the nucleus where they bind specific DNA elements and activate gene transcription.
  • JAKs are essential for multiple physiological functions. This has been demonstrated using genetically engineered mouse models that are deficient in specific JAKs. Jak1 -/- mice die perinatally, while Jak2 -/- mice have deficiencies in erythropoesis and die around day E12. Jak3 -/- mice are viable, but have a SCID phenotype with deficiencies in T cells, B cells and NK cells. TYK2 -/- mice exhibit features of hyper IgE syndrome. These phenotypes demonstrate the essential and non-redundant roles of JAK activity in vivo ( K. Ghoreschi, A. Laurence, J. J. O'Shea, Immunol. Rev. 228, 273 (2009 )).
  • JAK3 or the cognate common gamma chain cytokine receptor
  • J. J. O'Shea M. Pesu, D. C. Borie, P. S. Changelian, Nat. Rev. Drug Discov. 3, 555 (2004 )
  • Deletions of TYK2 result in hyper IgG syndrome and increased infection risk ( Y. Minegishi et al., Immunity. 25, 745 (2006 )).
  • No inactivating mutations have been reported for JAK1 or JAK2, consistent with the data from mice that demonstrates that JAK1 and JAK2 deficient mice are not viable.
  • JAK2 is the sole JAK family member involved in signal transduction of the critical hematopoetic cytokines IL-3, GMCSF, EPO and TPO.
  • JAK/STAT pathway link the JAK/STAT pathway to various diseases and disorders including hyperproliferative disorders and cancer such as leukemia and lymphomas, immunological and inflammatory disorders such as transplant rejection, asthma, chronic obstructive pulmonary disease, allergies, rheumatoid arthritis, type I diabetes, amyotropic lateral sclerosis and multiple sclerosis.
  • hyperproliferative disorders and cancer such as leukemia and lymphomas
  • immunological and inflammatory disorders such as transplant rejection, asthma, chronic obstructive pulmonary disease, allergies, rheumatoid arthritis, type I diabetes, amyotropic lateral sclerosis and multiple sclerosis.
  • the present invention provides novel compounds which are inhibitors of JAKs.
  • the invention also provides the novel compounds, as well as pharmaceutical compositions containing the compounds, for use in the treatment and prevention of JAK-mediated diseases.
  • the present invention provides compounds of formula I or pharmaceutically acceptable salts thereof:
  • Representative compounds of the instant invention include, but are not limited to the following compounds and their pharmaceutically acceptable salts thereof:
  • the invention also encompasses pharmaceutical compositions containing a compound of Formula I, and compounds of Formula I for use in treatment or prevention of JAK mediated diseases.
  • alkyl is intended to include both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbon atoms. Commonly used abbreviations for alkyl groups are used throughout the specification, e.g. methyl may be represented by “Me” or CH 3 , ethyl may be represented by “Et” or CH 2 CH 3 , propyl may be represented by “Pr” or CH 2 CH 2 CH 3 , butyl may be represented by "Bu” or CH 2 CH 2 CH 2 CH 3 , etc.
  • C 1-6 alkyl (or “C 1- C 6 alkyl”) for example, means linear or branched chain alkyl groups, including all isomers, having the specified number of carbon atoms.
  • C 1-6 alkyl includes all of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
  • C 1-4 alkyl means n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
  • alkoxy represents a linear or branched alkyl group of indicated number of carbon atoms attached through an oxygen bridge.
  • Acyl means a -C(O)R radical where R is optionally substituted alkyl, alkenyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, etc.
  • Acylamino means a -NRR' radical where R is H, OH, or alkoxy and R' is acyl, as defined herein.
  • alkyl refers to an aliphatic hydrocarbon group which may be straight or branched and having the indicated number of carbon atoms.
  • alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s - and t -butyl, pentyl, hexyl, and the like.
  • heteroalkyl refers to an alkyl group where 1, 2, or 3 of the carbon atoms is substituted by a heteroatom independently chosen from N, O, or S.
  • alkenyl refers to an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be straight or branched and having the indicated number of carbon atoms. Preferably alkenyl contains one carbon to carbon double bond, and up to four nonaromatic carbon-carbon double bonds may be present. Examples of alkenyl groups include ethenyl, propenyl, n-butenyl, 2-methyl-1-butenyl, 3-methylbut-2-enyl, n-pentenyl, octenyl and decenyl.
  • Alkynyl refers to an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond and which may be straight or branched and having the indicated number of carbon atoms.
  • suitable alkynyl groups include ethynyl, propynyl, 2-butynyl and 3-methylbutynyl.
  • Alkoxy refers to an alkyl-O- group in which the alkyl group is as described above.
  • C 1-6 alkoxy for example, includes methoxy, ethoxy, propoxy, isopropoxy, and the like.
  • Alkoxyalkyl refers to an alkyl group as described above in which one or more (in particular 1 to 3) hydrogen atoms have been replaced by alkoxy groups. Examples include CH 2 OCH 3 , CH 2 CH 2 OCH 3 and CH(OCH 3 )CH 3 .
  • Aminoalkyl refers to an alkyl group as described above in which one hydrogen atom has been replaced by an amino, monoalkylamino or dialkylamino group. Examples include CH 2 NH 2 , CH 2 CH 2 NHCH 3 and CH(N(CH 3 ) 2 )CH 3 .
  • C 0 as employed in expressions such as "C 0-6 alkyl" means a direct covalent bond; or when the term appears at the terminus of a substituent, C 0-6 alkyl means hydrogen or C 1-6 alkyl.
  • an integer defining the presence of a certain number of atoms in a group is equal to zero, it means that the atoms adjacent thereto are connected directly by a bond. For example, in the structure wherein s is an integer equal to zero, 1 or 2, the structure is when s is zero.
  • halogen refers to fluorine, chlorine, bromine and iodine (alternatively referred to as fluoro (F), chloro (Cl), bromo (Br), and iodo (I)).
  • aryl refers to aromatic mono- and poly-carbocyclic ring systems, wherein the individual carbocyclic rings in the polyring systems are fused or attached to each other via a single bond.
  • Suitable aryl groups include phenyl, naphthyl, 2,3-dihydro-1 H -indenyl, and biphenyl.
  • carbocycle (and variations thereof such as “carbocyclic” or “carbocyclyl”) as used herein, unless otherwise indicated, refers to (i) a C 3 to C 8 monocyclic, saturated or unsaturated ring or (ii) a C 7 to C 12 bicyclic saturated or unsaturated ring system. Each ring in (ii) is either independent of, or fused to, the other ring, and each ring is saturated or unsaturated.
  • the carbocycle may be attached to the rest of the molecule at any carbon atom which results in a stable compound.
  • fused bicyclic carbocycles are a subset of the carbocycles; i.e., the term "fused bicyclic carbocycle” generally refers to a C 7 to C 10 bicyclic ring system in which each ring is saturated or unsaturated and two adjacent carbon atoms are shared by each of the rings in the ring system.
  • a fused bicyclic carbocycle in which one ring is saturated and the other is saturated is a saturated bicyclic ring system.
  • a fused bicyclic carbocycle in which one ring is benzene and the other is saturated is an unsaturated bicyclic ring system.
  • a fused bicyclic carbocycle in which one ring is benzene and the other is unsaturated is an unsaturated ring system.
  • Saturated carbocyclic rings are also referred to as cycloalkyl rings, e.g., cyclopropyl, cyclobutyl, etc.
  • carbocycle is unsubstituted or substituted with C 1-6 alkyl, C 1-6 alkenyl, C 1-6 alkynyl, aryl, halogen, NH 2 or OH.
  • a subset of the fused bicyclic unsaturated carbocycles are those bicyclic carbocycles in which one ring is a benzene ring and the other ring is saturated or unsaturated, with attachment via any carbon atom that results in a stable compound. Representative examples of this subset include the following:
  • Cyanoalkyl refers to an alkyl group as described above in which one hydrogen atom has been replaced by a cyano group. Examples include CH 2 CN, CH 2 CH 2 CN and CH(CN)CH 3 .
  • Cycloalkyl means a carbocyclic ring system having 3 to 12 ring carbon atoms; said ring system may be (a) a monocyclic saturated carbocycle optionally fused to a benzene or a partially unsaturated carbocycle, or (b) a bicyclic saturated carbocycle.
  • the rings are fused across two adjacent ring carbon atoms (e.g., decalin), at one ring carbon atom (e.g., spiro[2.2]pentane), or are bridged groups (e.g., norbornane).
  • C 3-8 cycloalkyl means a cyclic ring of an alkane having three to eight total carbon atoms (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl).
  • C 3-7 cycloalkyl means a cyclic ring of an alkane having three to eight total carbon atoms (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl).
  • C 3-7 cycloalkyl "C 3-6 cycloalkyl"
  • C 5-7 cycloalkyl and the like have analogous meanings.
  • Haloalkyl refers to an alkyl group as described above wherein one or more (in particular 1 to 5) hydrogen atoms have been replaced by halogen atoms, with up to complete substitution of all hydrogen atoms with halo groups.
  • C 1-6 haloalkyl for example, includes -CF 3 , -CF 2 CF 3 , CHFCH 3 , and the like.
  • Heterocycle represents a monocyclic or bicyclic 3-12 membered ring system in which at least one ring is non-aromatic (saturated or partially unsaturated) and containing at least one heteroatom selected from O, S and N.
  • the second ring may be a heteroaryl, heterocycle or a saturated, partially unsaturated or aromatic carbocycle, and the point(s) of attachment to the rest of the molecule may be on either ring.
  • the rings may be fused across two adjacent ring atoms (e.g., quinoline), at one ring carbon atom (e.g., 1,4-dioxaspiro[4.5]decane), or may be bridged groups (e.g. 8-azabicyclo[3.2.1]octanyl,).
  • Heterocyclyl therefore includes heteroaryls, as well as dihydro and tetrathydro analogs thereof. Attachment of a heterocyclyl substituent can occur via a carbon atom or via a heteroatom.
  • heterocycles include, but are not limited to, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiamorpholinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, dihydroimidazolyl, dihydroindolyl, 1,2,3,4-tetrahydroisoquinolinyl, 5,6,7,8-tetrahydroimidazo[1,2- a ]pyrazine, 2,3-dihydrobenzofuranyl, benzo-1,4-dioxanyl, benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolin
  • Saturated heterocyclics form a subset of the heterocycles; i.e., the terms “saturated heterocyclic and (C 3-12 )heterocycloalkyl” generally refers to a heterocycle as defined above in which the entire ring system (whether mono- or poly-cyclic) is saturated.
  • saturated heterocyclic ring refers to a 4- to 8-membered saturated monocyclic ring or a stable 7- to 12-membered bicyclic ring system which consists of carbon atoms and one or more heteroatoms selected from N, O and S.
  • Representative examples include piperidinyl, piperazinyl, azepanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl (or tetrahydrofuranyl) .
  • Heteroaryl means an aromatic monocyclic or multicyclic ring system having 5 to 14 ring atoms, preferably 5 to 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination.
  • a nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide.
  • Non limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N-substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, pyrazolo[1,5-a]pyrimidinyl, imidazo[1,2-a]pyridinyl, imidazo[2,1-b]thiazolyl, indazolyl, benzofurazanyl, indolyl, azaindolyl, 20 benzimidazolyl, benzothienyl
  • bicyclic heterocycles include benzotriazolyl, indolyl, isoindolyl, indazolyl, indolinyl, isoindolinyl, quinoxalinyl, quinazolinyl, cinnolinyl, chromanyl, isochromanyl, tetrahydroquinolinyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, 2,3-dihydrobenzofuranyl, 2,3-dihydrobenzo-1,4-dioxinyl (i.e., ), imidazo(2,1- b )(1,3)thiazole, (i.e., ), and benzo-1,3-dioxolyl (i.e., ).
  • phenyl having as a substituent methylenedioxy attached to two adjacent carbon atoms.
  • Spirocyclyl or “spirocyclic ring” refers to a ring originating from a particular annular carbon of another ring.
  • a ring atom of a saturated bridged ring system (rings B and B'), but not a bridgehead atom, can be a shared atom between the saturated bridged ring system and a spirocyclyl (ring A) attached thereto.
  • a spirocyclyl can be carbocyclic or heteroalicyclic. In one embodiment, all rings of the spirocyclyl system are saturated. In another embodiement, the individual rings of the spirocyclyl system are selected from both saturated and unsaturated rings.
  • a heteroalicyclic, spirocyclyl, spiroheterocyclyl, or "spiroheterocyclic ring,” as used herein, refers to a bicyclic heterocyclic ring as defined above wherein the two rings are joined through a common ring carbon atom.
  • a spiroheterocyclic ring is a 3- to 12-membered ring system containing one to three heteroatoms, e.g., one to two heteroatoms, selected from the group consisting of N and O.
  • Non-limiting examples of spiroheterocyclyl, spiroheterocyclic rings include 2-oxa-6-azaspiro[3,3]heptane, 6-oxa-9-azaspiro[4.5]dec-9-yl, and 1,4-dioxaspiro[4.5]decane.
  • Non-limiting examples of a carbocyclic spirocyclyl systems comprising include: spiro[2.2]pentane, spiro[cylclobutane-1,2'-indene], spiro[4.4]nonane, and spiro[4.5]decane.
  • Hydroalkyl refers to an alkyl group as described above in which one or more (in particular 1 to 3) hydrogen atoms have been replaced by hydroxy groups. Examples include CH 2 OH, CH 2 CHOH and CHOHCH 3 .
  • Alkylene alkenylene, alkynylene, cycloalkylene,” “arylene,” “heteroarylene,” and “heterocyclylene” refer to a divalent radical obtained by the removal of one hydrogen atom from an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl group, respectively, each of which is as defined above.
  • an “unsaturated” ring is a partially or fully unsaturated ring.
  • an “unsaturated monocyclic C 6 carbocycle” refers to cyclohexene, cyclohexadiene, and benzene.
  • heterocycle described as containing from “ 1 to 4 heteroatoms” means the heterocycle can contain 1, 2, 3 or 4 heteroatoms.
  • sulfamoyl is a suffix to denote radicals derived from sulfamide such as -SO 2 NH 2 , -SO 2 NHR and -SO 2 N(RR 1 ).
  • substituted e.g., as in "aryl which is optionally substituted with one or more substituents "
  • substituents include mono- and poly-substitution by a named substituent to the extent such single and multiple substitution (including multiple substitution at the same site) is chemically allowed.
  • oxy means an oxygen (O) atom.
  • thio means a sulfur (S) atom.
  • any variable e.g., R 2 , R 3 , etc.
  • its definition in each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups can be on the same carbon or on different carbons, so long as a stable structure results.
  • the phrase "optionally substituted with one or more substituents” should be taken to be equivalent to the phrase “optionally substituted with at least one substituent” and in such cases one embodiment will have from zero to three substituents.
  • Structural representations of compounds having substituents terminating with a methyl group may display the terminal methyl group either using the characters "CH 3 ", e.g. "-CH 3 " or using a straight line representing the presence of the methyl group, e.g. " - ", i.e., have equivalent meanings.
  • variable definitions containing terms having repeated terms e.g., (CR i Rj) r , where r is the integer 2, R i is a defined variable, and Rj is a defined variable
  • the value of R i may differ in each instance in which it occurs
  • the value of Rj may differ in each instance in which it occurs.
  • R i and Rj are independently selected from the group consisting of methyl, ethyl, propyl and butyl, then (CR i Rj) 2 can be
  • Patient includes both human and animals.
  • “Mammal” means humans and other mammalian animals.
  • “Therapeutically effective amount” means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, a system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • treatment includes alleviating, ameliorating, relieving or otherwise reducing the signs and symptoms associated with a disease or disorder.
  • composition as in pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) (pharmaceutically acceptable excipients) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of Formula I, and pharmaceutically acceptable excipients.
  • optionally substituted means "unsubstituted or substituted," and therefore, the generic structural formulas described herein encompasses compounds containing the specified optional substituent as well as compounds that do not contain the optional substituent.
  • A is selected from: isoindolinyldiyl, pyridinyldiyl, and indolinyldiyl.
  • A is selected from:
  • A is selected from pyridinyldiyl
  • R 1 is selected from hydrogen, C 1-2 alkyl, fluoro, and hydroxy. In a variant of this embodiment, R 1 is hydrogen or fluoro. In yet another embodiment R 1 is hydrogen.
  • n is 0 or 1. In another embodiment, m is 2.
  • R 2 is selected from:
  • R 2 is selected from:
  • R 2 is selected from:
  • R 2 is selected from: carboxy, (8-oxa-3-azabicyclo[3.2.1]oxtane)carbonyl, cyclohexyl, piperidinyl, morpholinylcarbonyl, azepanyl, ((1 R ,4 R )-2-oxa-5-azabicyclo[2.2.1]heptyl)carbonyl, cyanomethyl, (2-oxa-6-azaspiro[3.3]heptyl)carbonyl, thiomorpholinylcarbonyl, ((1 R ,4 R )-2-oxa-5-azabicyclo[2.2.1]heptyl)carbonyl, (2-oxa-5-azabicyclo[2.2.1]heptyl)carbonyl, piperidinylcarbonyl pyrrolidinylcarbonyl, pyrrolidinyl, tert -butylaminomethyl, 2,2,2-trifluoroethyl, pyr
  • each R 3 is independently selected from: C 1-10 alkyl, oxo, (C 3-12 )heterocycloalkyl C 0-10 alkyl, C 1-10 alkoxyC 0-10 alkyl, halogen, and C 1-6 haloalkyl.
  • each R 3 is independently selected from: methyl, trifluoromethyl, ethyl, trifluoroethyl, fluoro, oxo, hydroxy, isopropyl, thiomorpholinyl, isobutyl, and difluoromethyl.
  • each R 4 is independently selected from: C 1- 10 alkyl, (C 3-12 )heterocycloalkyl C 0-10 alkyl, C 1-10 alkoxyC 0-10 alkyl, C 0-10 alkylaminoC 0-10 alkyl, halogen, hydroxy, C 1-6 haloalkyl, and oxo.
  • R 4 independently is selected from: methyl, fluoro, 2,2,2-trifluoroethyl, trifluoromethyl, tert -butylamino, methoxy, hydroxy, oxo, isopropyl, thiomorpholinyl, and isobutyl.
  • Compounds of Formula I contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers.
  • the present invention is meant to comprehend all such isomeric forms of the compounds of Formula I, either as single species or mixtures thereof.
  • tautomers Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. Such an example may be a ketone and its enol form known as keto-enol tautomers. The individual tautomers as well as mixture thereof are encompassed with compounds of Formula I.
  • Specific embodiments of the present invention include a compound which is selected from the group consisting of the subject compounds of the examples herein or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention may contain one or more asymmetric centers and can thus occur as "stereoisomers” including racemates and racemic mixtures, enantiomeric mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within the scope of this invention. The present invention is meant to comprehend all such isomeric forms of these compounds.
  • the compounds of Formula (I) may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of Formula (I) as well as mixtures thereof, including racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric isomers. For example, if a compound of Formula (I) incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the invention.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • some of the compounds of Formula (I) may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
  • Enantiomers can also be separated by use of chiral HPLC column
  • All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds including those of the salts, solvates and esters of the compounds), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention.
  • enantiomeric forms which may exist even in the absence of asymmetric carbons
  • rotameric forms which may exist even in the absence of asymmetric carbons
  • atropisomers rotameric forms
  • diastereomeric forms are contemplated within the scope of this invention.
  • a compound of Formula (I) incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the invention.
  • keto-enol and imine-enamine forms of the compounds are included in the invention.
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 recommendations.
  • the use of the terms "salt”, “solvate”, “ester” and the like, is intended to equally apply to the salt, solvate and ester of enantiomers, stereoisomers, rotamers, tautomers for racemates of the inventive compounds.
  • stereomeric nomenclature includes "or", for example, tert -butyl 4-(3-(4-(( 3S,5S or 3 R ,5 R )-3,5-dimethylmorpholine-4-carbonyl)-3-methylphenylamino)-4-oxo-4,5-dihydropyrazolo[4,3- c ]pyridin-1-yl)-4-ethylpiperidine-1-carboxylate, the "or” indicates that chiral resolution of racemate into individual enantiomers was accomplished but the actual optical activity of the specific enantiomer was not necessarily determined.
  • the coupling reaction is often the formation of salts using an enantiomerically pure acid or base.
  • the diasteromeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue.
  • the racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art.
  • any enantiomer of a compound can be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N , N '-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N -ethyl-morpholine, N -ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, 1-hydroxy-2-naphthoic acid (xinafoate) and the like.
  • Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, xinafoate and tartaric acids.
  • references to the compound of Formula I subsets thereof, embodiments thereof, as well as specific compounds are meant to also include the pharmaceutically acceptable salts and stereoisomers thereof
  • crystalline forms for compounds of the present invention may exist as polymorphs and as such all forms are intended to be included in the present invention.
  • some of the compounds of the instant invention may form solvates with water (hydrates) or common organic solvents. Such solvates are encompassed within the scope of this invention.
  • the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature.
  • the present invention is meant to include all suitable isotopic variations of the compounds of generic Formula I.
  • different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H).
  • Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
  • Isotopically-enriched compounds within generic Formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the schemes and examples herein using appropriate isotopically-enriched reagents and/or intermediates.
  • Compound of Formula I or its pharmaceutically acceptable salts and pharmaceutical compositions are for use to treat or prevent a variety of conditions or diseases mediated by Janus kinases, in particular diseases or conditions that can be ameliorated by the inhibition of a Janus kinase such as JAK1, JAK2, JAK3 or TYK2.
  • Such conditions and diseases include, but are not limited to:
  • Another aspect of the present invention provides for a compound of Formula I for use in the treatment or prevention of a JAK-mediated disease or disorder.
  • One aspect of the invention is a compound of Formula I or a pharmaceutically acceptable salt or a stereoisomer thereof for use in the treatment of a disease or a disorder ameliorated by inhibition of Janus kinases JAK1 and JAK2.
  • Another aspect of the invention is a compound of Formula I or a pharmaceutically acceptable salt or a stereoisomer thereof and a second active agent for use in the treatment of a disease or a disorder ameliorated by inhibition of Janus kinases JAK1 and JAK2.
  • prophylactic or therapeutic dose of a compound of Formula I will, of course, vary with the nature and the severity of the condition to be treated and with the particular compound of Formula I and its route of administration. It will also vary according to a variety of factors including the age, weight, general health, sex, diet, time of administration, rate of excretion, drug combination and response of the individual patient. In general, the daily dose from about 0.001 mg to about 100 mg per kg body weight of a mammal, preferably 0.01 mg to about 10 mg per kg. On the other hand, it may be necessary to use dosages outside these limits in some cases.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a formulation intended for the oral administration of humans may contain from 0.05 mg to 5 g, of active agent compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 99.95 percent of the total composition.
  • the dosage unit forms may contain from about 0.05 to about 3g of active ingredient.
  • Dosage unit forms will generally contain between from about 0.1 mg to about 0.4 g of an active ingredient, typically 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 1 mg, 2 mg, 5 mg, 10 mg, 25 mg, 50 mg, 100 mg, 200 mg, or 400 mg.
  • compositions comprising a compound of Formula I with a pharmaceutically acceptable carrier.
  • compounds of Formula I may be administered orally, by inhalation spray, topically, parenterally or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques.
  • the compound of the invention is effective in the treatment of humans.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Patent 4,256,108 ; 4,166,452 ; and 4,265,874 to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredients is mixed with water-miscible solvents such as propylene glycol, PEGs and ethanol, or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water-miscible solvents such as propylene glycol, PEGs and ethanol
  • an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoo
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • flavoring agents such as sucrose, saccharin or aspartame.
  • sweetening agents such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerin, glycerin, glycerin, glycerin, glycerin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. Cosolvents such as ethanol, propylene glycol or polyethylene glycols may also be used.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Dosage forms for inhaled administration may conveniently be formulated as aerosols or dry powders.
  • the active substance is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronization.
  • the medicinal preparation is adapted for use with a pressurized metered dose inhaler (pMDI) which releases a metered dose of medicine upon each actuation.
  • pMDI pressurized metered dose inhaler
  • the formulation for pMDIs can be in the form of solutions or suspensions in halogenated hydrocarbon propellants.
  • the type of propellant being used in pMDIs is being shifted to hydrofluoroalkanes (HFAs), also known as hydrofluorocarbons (HFCs).
  • HFAs hydrofluoroalkanes
  • HFCs hydrofluorocarbons
  • 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoropropane (HFA 227) are used in several currently marketed pharmaceutical inhalation products.
  • the composition may include other pharmaceutically acceptable excipients for inhalation use such as ethanol, oleic acid, polyvinylpyrrolidone
  • Pressurized MDIs typically have two components. Firstly, there is a canister component in which the drug particles are stored under pressure in a suspension or solution form. Secondly, there is a receptacle component used to hold and actuate the canister. Typically, a canister will contain multiple doses of the formulation, although it is possible to have single dose canisters as well.
  • the canister component typically includes a valve outlet from which the contents of the canister can be discharged.
  • Aerosol medication is dispensed from the pMDI by applying a force on the canister component to push it into the receptacle component thereby opening the valve outlet and causing the medication particles to be conveyed from the valve outlet through the receptacle component and discharged from an outlet of the receptacle.
  • the medication particles are "atomized", forming an aerosol. It is intended that the patient coordinate the discharge of aerosolized medication with his or her inhalation, so that the medication particles are entrained in the patient's aspiratory flow and conveyed to the lungs.
  • pMDIs use propellants to pressurize the contents of the canister and to propel the medication particles out of the outlet of the receptacle component.
  • the formulation is provided in a liquid or suspension form, and resides within the container along with the propellant.
  • the propellant can take a variety of forms.
  • the propellant can comprise a compressed gas or liquefied gas.
  • the medicinal preparation is adapted for use with a dry powder inhaler (DPI).
  • DPI dry powder inhaler
  • the inhalation composition suitable for use in DPIs typically comprises particles of the active ingredient and particles of a pharmaceutically acceptable carrier.
  • the particle size of the active material may vary from about 0.1 ⁇ m to about 10 ⁇ m; however, for effective delivery to the distal lung, at least 95 percent of the active agent particles are 5 ⁇ m or smaller.
  • Each of the active agent can be present in a concentration of 0.01 - 99%. Typically however, each of the active agents is present in a concentration of about 0.05 to 50%, more typically about 0.2 - 20% of the total weight of the composition.
  • the inhalable powder preferably includes pharmaceutically acceptable carrier, which may be composed of any pharmacologically inert material or combination of materials which is acceptable for inhalation.
  • the carrier particles are composed of one or more crystalline sugars; the carrier particles may be composed of one or more sugar alcohols or polyols.
  • the carrier particles are particles of dextrose or lactose, especially lactose.
  • the particle size of the carrier particles may range from about 10 microns to about 1000 microns.
  • the particle size of the carrier particles may range from about 20 microns to about 120 microns. In certain other embodiments, the size of at least 90% by weight of the carrier particles is less than 1000 microns and preferably lies between 60 microns and 1000 microns. The relatively large size of these carrier particles gives good flow and entrainment characteristics. Where present, the amount of carrier particles will generally be up to 95%, for example, up to 90%, advantageously up to 80% and preferably up to 50% by weight based on the total weight of the powder. The amount of any fine excipient material, if present, may be up to 50% and advantageously up to 30%, especially up to 20%, by weight, based on the total weight of the powder.
  • the powder may optionally contain a performance modifier such as L-leucine or another amino acid, and/or metals salts of stearic acid such as magnesium or calcium stearate.
  • Compounds of Formula I may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ambient temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ambient temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • Topical formulations may generally be comprised of a pharmaceutical carrier, cosolvent, emulsifier, penetration enhancer, preservative system, and emollient.
  • compound of Formula I may be co-administered with other therapeutic agents.
  • the present invention provides pharmaceutical compositions for treating JAK mediated diseases comprising a therapeutically effective amount of a compound of Formula I and one or more other therapeutic agents.
  • a compound of Formula I may be combined with agents such as: (1) TNF- ⁇ inhibitors such as Remicade® and Enbrel®); (2) non-selective COX-I/COX-2 inhibitors (such as piroxicam, diclofenac, propionic acids such as naproxen, flubiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, apazone, pyrazolones such as phenylbutazone, salicylates such as aspirin); (3) COX-2 inhibitors (such as meloxicam, celecoxib, rofecoxib, valdecoxib and etoricoxib); (4) other agents for treatment of rheuma
  • TNF- ⁇ inhibitors such as Remicade® and Enbrel®
  • COX-I/COX-2 inhibitors such as piroxicam, diclofenac, prop
  • the compounds of the present invention can be prepared according to the following general schemes using appropriate materials, and are further exemplified by the subsequent specific examples.
  • the compounds illustrated in the examples are not to be construed as forming the only genus that is considered as the invention.
  • the illustrative Examples below, therefore, are not limited by the compounds listed or by any particular substituents employed for illustrative purposes. Substituent numbering as shown in the schemes does not necessarily correlate to that used in the claims and often, for clarity, a single substituent is shown attached to the compound where multiple substituents are allowed under the definitions of the instant invention herein above.
  • This coupling could be carried out using an appropriate catalytic palladium-ligand system, such as Pd 2 (dba) 3 or Pd 2 (dba) 3 ⁇ CHCl 3 , and 2-di- tert- butylphosphino-2',4',6'-triisopropylbiphenyl ( t -Bu XPhos) or di- tert -butyl[3,4,5,6-tetramethyl-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane (Me 4 t Bu-XPhos), or 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (XPhos).
  • an appropriate catalytic palladium-ligand system such as Pd 2 (dba) 3 or Pd 2 (dba) 3 ⁇ CHCl 3
  • this cross coupling is carried out using either 2-propanol or t-amyl alcohol solvents, employing either KOAc or K 3 PO 4 base at between 70-80°C.
  • Reduction of the nitrile to aldehyde IV could then be carried out with DIBAL-H, at or between - 78°C and 0°C in a suitable solvent such as DCM.
  • Further reduction of the aldehyde could be accomplished via a two step process via dithiane formation using HSCH 2 CH 2 SH in the presence of acid, followed by reduction with Raney Ni to afford the ethyl substituted piperidine V.
  • This coupling could be carried out using an appropriate catalytic palladium-ligand system, such as Pd 2 (dba) 3 or Pd 2 (dba) 3 ⁇ CHCl 3 , and 2-di- tert -butylphosphino-2',4',6'-triisopropylbiphenyl ( t -Bu XPhos) or di- tert -butyl[3,4,5,6-tetramethyl-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane (Me 4 t Bu-XPhos), or 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (XPhos).
  • Step 2 4-(benzyloxy)-1 H -pyrazolo[4,3- c ]pyridin-3-amine ( I-1 )
  • Step 1 tert-butyl 4-(3-amino-4-(benzyloxy)-1 H -pyrazolo [4,3- c ]pyridin-1-yl)-4-(cyanomethyl)piperidme-1-carboxylate
  • Step 2 tert -butyl 4- ⁇ 4-(benzyloxy)-3-[bis( tert -butoxycarbonyl)amino]-1 H -pyrazolo[4,3- c ]pyridin-1-yl ⁇ -4-(cyanomethyl)piperidine-1-carboxylate
  • Step 3 tert -butyl 4- ⁇ 4-(benzyloxy)-3-[bis( tert -butoxycarbopyl)amino]-1 H -pyrazolo[4,3- c ]pyridin-1-yl ⁇ -4-(2-oxoethyl)piperidine-1-carboxylate
  • Bis(cyclopentadienyl)zirconium chloride hydride (3.11 g, 12.1 mmol) was added in portions to a stirred, ambient temperature solution of tert -butyl 4- ⁇ 4-(benzyloxy)-3-[bis( tert- butoxycarbonyl)amino]-1 H- pyrazolo[4,3-c]pyridin-1-yl ⁇ -4-(cyanomethyl)piperidine-1-carboxylate (4.00 g, 6.04 mmol) in DCM (60 mL). The solution was stirred at ambient temperature for 10 minutes, then the reaction was quenched with water (400 mL) extracted with DCM ( ⁇ 3).
  • Step 4 tert -butyl 4- ⁇ 4-(benzyloxy)-3-[bis( tert -butoxycarbonyl)amino]-1 H -pyrazolo[4,3- c ]pyridin-1-yl ⁇ -4-(2-hydroxyethyl)piperidine-1-carboxylate
  • Step 5 tert-butyl 4- ⁇ 4-(benzyloxy)-3-[bis( tert -butoxycarbonyl)amino]-1 H -pyrazolo[4,3- c ]pyridin-1-yl ⁇ -4-(2-methane sulfonyl ethyl)piperidine-1-carboxylate
  • Step 6 tert -butyl 4-(4-(benzyloxy)-3-(( tert -butoxycarbonyl)amino1 H -pyrazolo[4,3- c ]pyridin-1-yl)-4-ethylpiperidine-1-carboxylate
  • Step 7 3-amino-1-(4-ethylpiperidin-4-yl)-1 H -pyrazolo[4,3- c ]pyridin-4(5 H )-one hydrochloride salt
  • Step 8 tert -butyl 4-(3-amino-4-oxo-4,5-dihydro-1 H -pyrazolo[4,3- c ]pyridin-1-yl)-4-ethylpiperidine-1-carboxylate ( I-3 )
  • Step 1 ( R or S ) 2-(4-bromophenyl)-1,1,1-trifluoropropan-2-ol ( I-5 )
  • Step 2 ( R or S ) 1 -bromo-4-(1,1,1-trifluoro-2-methoxypropan-2-yl)benzene ( I-6 )
  • I-6A was prepared in an analogous manner to I-6A above, using I-5B .
  • Step 2 4-bromo- N -( tert -butyl)-2-methylbenzenesulfonamide
  • Step 3 5-bromo-2-( tert -butyl)benzo[ d ]isothiazol-3(2 H )-one-1,1-dioxide
  • Step 4 5-bromo-2-( tert -butyl)-2,3-dihydrobenzo[d]isothiazole-1,1-dioxide ( I-8 )
  • Step 1 4-bromo-2-methyl- N -(tetrahydro-2 H -pyran-4-yl)benzenesulfonamide
  • Step 2 4-bromo-2-(bromomethyl)- N -(tetrahydro-2 H -pyran-4-yl)benzenesulfonamide
  • Step 3 5-bromo-2-(tetrahydro-2 H -pyran-4-yl)-2,3-dihydrobenzo[ d ]isothiazole 1,1-dioxide ( I-9 )
  • Step 1 1-(4-bromophenyl)-2,2,2-trifluoroethyl trifluoromethanesulfonate
  • Step 2 N -(1-(4-bromophenyl)-2,2,2-trifluoroethyl)-2-methylpropan-2-amine ( I-11 )
  • 1-(4-Bromophenyl)-2,2,2-trifluoroethyl trifluoromethanesulfonate (7.59 g, 19.6 mmol) was dissolved in cyclohexane (70 mL) and 2-methylpropan-2-amine (6.23 mL, 58.8 mmol), DMAP (0.240 g, 1.96 mmol), and ground, dried potassium carbonate (5.42 g, 39.2 mmol) (dried over vacuum at 60°C for one hour) was added. The reaction mixture was heated to 75°C and stirred for 48 hours. The reaction mixture was diluted with DCM and washed with water.
  • Step 2 tert -butyl 3-4-bromobenzoyl)-2-oxopiperidine-1-carboxylate
  • Lithium bis(trimethylsilyl)amide (2.11 mL, 2.11 mmol, 1.0 M in THF) was added to a solution of tert -butyl 2-oxopiperidine-1-carboxylate (0.20 g, 1.0 mmol) in THF (2 mL) at - 78°C. The resulting mixture was stirred for 10 minutes, then 4-bromobenzoyl chloride (0.22 g, 1.0 mmol) was added. The reaction was warmed to ambient temperature and stirred for 1 hour before being quenched with saturated aqueous ammonium chloride (20 mL).
  • Step 4 ( R or S )-2-(4-bromophenyl)-2-(trifluoromethyl)piperidine ( I-12A & I-12B )
  • Step 3 ( S or R )-2-(4-bromophenyl)-2-(trifluoromethyl)pyrrolidine ( I-13A & I-13B )
  • Step 1 tert -butyl 3-(4-bromobenzoyl)-2-oxoazepane-1-carboxylate
  • Step 4 ( R or S )-2-(4-bromophenyl)-2-(trifluoromethyl)azepane and ( S )-2-(4-bromophenyl)-2-(trifluoromethyl)azepane ( I-14A & I-14B )
  • Table 2 discloses Examples that were prepared in analogy to 1-15-1.
  • Table 2. Example Structure Compound Name LRMS I-15-2 (4-bromo-2-methylphenyl)(morpholino)methanone LRMS (ESI) Calc'd for C 12 H 15 BrNO 2 [M+H] + : 286, found 286. I-15-3 (4-bromo-2-methylphenyl)(thiomorpholino) methanone LRMS (ESI) Calc'd for C 12 H 15 BrNOS [M+H] + : 302, found 302.
  • Step 2 8-oxa-3-azabicyclo[3.2.1]octan-3-yl(4-bromo-2-methylphenyl)methanone ( I-16-1 )
  • Table 3 discloses intermediates that were prepared in an analogous manner to that of I-16-1 above.
  • I-16-15A and I-16-15B trans (4-bromo-2-methylphenyl) (3,5-dimethylmorpholino) methanone Racemic aryl bromide was separated by prep HPLC using a Chiralcel OJ-H, eluting with 10% EtOH in hexanes, retention times 7.5 and 10.0 minutes.
  • Step 2 tert -butyl 4-(4-bromo-2-(bromomethyl)phenylsulfonamido)piperidine-1-carboxylate
  • Step 3 tert-butyl 4-(5-bromo-1,1-dioxidobenzo[ d ]isothiazol-2(3 H )-yl)piperidine-1-carboxylate
  • Step 4 5-bromo-2-(piperidin-4-yl)-2,3-dihydrobenzo[ d ]isothiazole 1,1-dioxide
  • Step 5 5-bromo-2-(1-methylpiperidin-4-yl)-2,3-dihydrobenzo[d]isothiazole 1,1-dioxide ( I-18 )
  • Step 3 5-bromo-2-(4,4-difluoro-1-methylcyclohexyl)isoindolin-1-one ( I-19 )
  • Triethylamine (3.39 g, 33.5 mmol) was added to methyl 4-bromo-2-(bromomethyl)benzoate (2.06 g, 6.70 mmol) and 4,4-difluoro-1-methylcyclohexanamine (1.00 g, 6.70 mmol) in toluene (60 mL). The mixture was stirred at 110°C for 16 hours, then concentracted in vacuo and purified by silica chromatography, eluting with 0-6% EtOAc/hexanes to afford the title compound as a solid.
  • reaction mixture was quenched with water (20 mL), extracted with ethyl acetate ( ⁇ 3), and the combined organic layers were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated in vacuo.
  • the residue was purified by silica chromatography, eluting with 0-10% DCM/MeOH to afford the title compound as a solid.
  • Step 1 tert -butyl 4-(4-(benzyloxy)-3-((4-( N , N -dimethylsulfamoyl)phenyl)amino)-1 H- pyrazolo[4,3- c ]pyridin-1-yl)-4-(cyanomethyl)piperidine-1-carboxylate
  • Step 2 tert -butyl 4-(4-(benzyloxy)-3-((4-( N , N -dimethylsulfamoyl)phenyl)amino)-1 H- pyrazolo[4,3- c ]pyridin-1-yl)-4-(2-oxoethyl)piperidine-1-carboxylate
  • Step 3 tert -butyl 4-((1,3-dithiolan-2-yl)methyl)-4-(3-((4-( N , N -dimethylsulfamoyl)phepyl) amino)-4-oxo-4,5-dihydro-1 H -pyrazolo[4,3- c ]pyridin-1-yl)piperidine-1-carboxylate
  • Step 4 tert-butyl 4-(3-(4-( N , N -dimethylsulfamoyl)phenylamino)-4-oxo-4,5-dihydropyrazolo[4,3- c ]pyridin-1-yl)-4-ethylpiperidine-1-carboxylate
  • Tris(dibenzylideneacetone) dipalladium(0)-chloroform adduct 34 mg, 0.033 mmol was added to tert -butyl 4-(3-amino-4-oxo-4,5-dihydro-1 H -pyrazolo[4,3- c ]pyridin-1-yl)-4-ethylpiperidine-1-carboxylate (0.12 g, 0.33 mmol), 8-oxa-3-azabicyclo[3.2.1]octan-3-yl(4-bromo-2-methylphenyl)methanone (0.12 g, 0.40 mmol), dibutyl(2',4',6'-triisopropyl-[1,1'-biphenyl]-2-yl)phosphine (28 mg, 0.066 mmol) and potassium acetate (65 mg, 0.66 mmol) in 2-propanol (1 mL).
  • Table 5 discloses intermediates that were prepared in an analogous manner to that of Example 2-1 above.
  • the reaction could alternatively employ t BuXPhos-Pd-G3, [(2-Di- tert -butylphosphino-2',4',6'-triisopropyl-1,1'-biphenyl)-2-(2'-amino-1,1'-biphenyl)] palladium(II) methanesulfonate, or XantPhos ligand in place of the t-BuXPhos ligand, in t -amyl alcohol, at or around 70-80°C. Table 5.
  • Morpholin-2-one (29 mg, 0.28 mmol), 4-methylmorpholine (29 mg, 0.28 mmol) and ((1 H -benzo[ d ][1,2,3]triazol-1-yl)oxy)tri(pyrrolidin-1-yl)phosphonium (PyBop) (0.10 g, 0.28 mmol) were added to a stirred solution of 4-((1-(1-( tert -butoxycarbonyl)-4-ethylpiperidin-4-yl)-4-oxo-4,5-dihydro-1 H -pyrazolo[4,3- c ]pyridin-3-yl)amino)-2-methylbenzoic acid (70 mg, 0.14 mmol) in DCM (10 mL) at ambient temperature.
  • the reaction mixture was stirred for 12 hours at ambient temperature, then quenched with water (5 mL) and extracted with EtOAc ( ⁇ 2). The combined organic layers were washed with brine ( ⁇ 2), dried over anhydrous Na 2 SO 4 , filtered, and the filtrate was concentrated in vacuo.
  • the product was purified by Prep-HPLC using an Xbridge C18 column, eluting with acetonitrile in water (containing 0.05% NH 4 HCO 3 ) using a acetonitrile gradient of 20% acetonitrile hold for 6 minutes, then hold at 54% for 2 minutes, down to 20% in 2 minutes. The collected fractions were combined and concentrated in vacuo to afford the title compound as a solid.
  • the ability of compounds to inhibit the activity of JAK1, JAK2, JAK3, and Tyk2 was measured using a recombinant purified GST-tagged catalytic domain for each enzyme (Invitrogen JAK1 #M4290, JAK2 #M4290, JAK3 #M4290, Tyk2 #M4290) in an HTRF format biochemical assay.
  • the reactions employed a common peptide substrate, LCB-EQEDEPEGDYFEWLW-NH2 (in-house).
  • the basic assay protocol is as follows: First, 250 nL of diluted compounds in DMSO were dispensed into the wells of a dry 384-well Black plate (Greiner #781076) using a Labcyte Echo 555 acoustic dispenser.
  • reagent additions employed an Agilent Bravo.
  • 18 ⁇ L of 1.11X enzyme and 1.11X substrate in IX assay buffer (Invitrogen kinase buffer # PV3189, 2 mM DTT, 0.05% BSA) were added to the wells and shaken and then preincubated for 30 minutes at room temperature to allow compound binding to equilibrate.
  • 2 ⁇ L of 10X ATP in IX assay buffer was added to initiate the kinase reaction and the plates were shaken and then incubated at room temperature for 120 minutes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
EP15853280.4A 2014-10-22 2015-10-21 Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors Active EP3209296B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/CN2014/089139 WO2016061751A1 (en) 2014-10-22 2014-10-22 Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors
PCT/US2015/056541 WO2016064935A1 (en) 2014-10-22 2015-10-21 Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors

Publications (3)

Publication Number Publication Date
EP3209296A1 EP3209296A1 (en) 2017-08-30
EP3209296A4 EP3209296A4 (en) 2018-04-11
EP3209296B1 true EP3209296B1 (en) 2019-07-10

Family

ID=55760043

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15853280.4A Active EP3209296B1 (en) 2014-10-22 2015-10-21 Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors

Country Status (11)

Country Link
US (1) US10072025B2 (hr)
EP (1) EP3209296B1 (hr)
JP (1) JP2017531672A (hr)
KR (1) KR20170068585A (hr)
CN (1) CN107106547A (hr)
AU (1) AU2015336035A1 (hr)
BR (1) BR112017008103A2 (hr)
CA (1) CA2964375A1 (hr)
MX (1) MX2017005281A (hr)
RU (1) RU2017117248A (hr)
WO (2) WO2016061751A1 (hr)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7034942B2 (ja) 2016-05-05 2022-03-14 エフ.ホフマン-ラ ロシュ アーゲー ピラゾール誘導体、その組成物及び治療的使用
CN106749081B (zh) * 2016-11-21 2019-06-25 浙江嘉圣生物医药有限公司 一种(s)-(4-溴-2-甲苯)(3-甲基吗啉)甲酮的制备方法
WO2018118947A1 (en) 2016-12-21 2018-06-28 Biotheryx, Inc. Thienopyrrole derivatives for use in targeting proteins, compositions, methods, and uses thereof
US10406165B2 (en) 2017-03-14 2019-09-10 Biotheryx, Inc. Compounds targeting proteins, compositions, methods, and uses thereof
HUE057748T2 (hu) 2017-12-22 2022-06-28 Siegfried Ag Egyedényes eljárás racém nikotin elõállítására, etil-nikotinát vinilpirrolidonnal végzett reakciójával, nátrium-etanolát vagy kálium-etanolát vagy ezek keverékeinek jelenlétében
CN111819181A (zh) * 2018-05-10 2020-10-23 罗欣药业(上海)有限公司 苯并七元杂环类化合物、其制备方法、药物组合物及应用
JP2022501401A (ja) * 2018-09-27 2022-01-06 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 自己免疫疾患の治療のためのヘテロシクリル化合物
CN114901659A (zh) 2019-11-26 2022-08-12 施万生物制药研发Ip有限责任公司 作为jak抑制剂的稠合嘧啶吡啶酮化合物
CN114939349B (zh) * 2022-05-03 2023-08-01 北京工业大学 一种用于气体分离的金属-有机空穴配合物混合基质膜的制备

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA111854C2 (uk) * 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн Способи і проміжні сполуки для отримання інгібіторів jak
CA2901766A1 (en) 2013-03-19 2014-09-25 Merck Sharp & Dohme Corp. N-(2-cyano heterocyclyl)pyrazolo pyridones as janus kinase inhibitors
WO2014146249A1 (en) * 2013-03-19 2014-09-25 Merck Sharp & Dohme Corp. Geminally substituted cyanoethylpyrazolo pyridones as janus kinase inhibitors
US10059705B2 (en) 2013-03-19 2018-08-28 Merck Sharp & Dohme Corp. Acyclic cyanoethylpyrazolo pyridones as janus kinase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
EP3209296A4 (en) 2018-04-11
US20170240567A1 (en) 2017-08-24
MX2017005281A (es) 2017-08-15
RU2017117248A (ru) 2018-11-22
WO2016061751A1 (en) 2016-04-28
KR20170068585A (ko) 2017-06-19
AU2015336035A1 (en) 2017-04-20
WO2016064935A1 (en) 2016-04-28
BR112017008103A2 (pt) 2018-02-20
CN107106547A (zh) 2017-08-29
EP3209296A1 (en) 2017-08-30
JP2017531672A (ja) 2017-10-26
CA2964375A1 (en) 2016-04-28
US10072025B2 (en) 2018-09-11

Similar Documents

Publication Publication Date Title
EP3209296B1 (en) Ethyl n-boc piperidinyl pyrazolo pyridones as janus kinase inhibitors
AU2012312305B2 (en) Acyclic cyanoethylpyrazoles as janus kinase inhibitors
US9725445B2 (en) Cycloalkyl nitrile pyrazolo pyridones as Janus kinase inhibitors
AU2014234909B2 (en) Acyclic cyanoethylpyrazolo pyridones as Janus kinase inhibitors
US9957265B2 (en) N-(2-cyano heterocyclyl) pyrazolo pyridones as janus kinase inhibitors
AU2014234907B2 (en) Geminally substituted cyanoethylpyrazolo pyridones as Janus kinase inhibitors
KR20150130310A (ko) 야누스 키나제 억제제로서의 같은자리 치환된 시아노에틸피라졸로 피리돈

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20170522

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602015033729

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0031437000

Ipc: C07D0471040000

A4 Supplementary search report drawn up and despatched

Effective date: 20180312

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 19/02 20060101ALI20180306BHEP

Ipc: A61K 31/541 20060101ALI20180306BHEP

Ipc: A61P 11/06 20060101ALI20180306BHEP

Ipc: A61K 31/4545 20060101ALI20180306BHEP

Ipc: A61K 31/554 20060101ALI20180306BHEP

Ipc: C07D 519/00 20060101ALI20180306BHEP

Ipc: A61K 31/538 20060101ALI20180306BHEP

Ipc: C07D 471/04 20060101AFI20180306BHEP

Ipc: A61K 31/553 20060101ALI20180306BHEP

Ipc: A61K 31/5377 20060101ALI20180306BHEP

Ipc: A61P 35/00 20060101ALI20180306BHEP

Ipc: A61K 31/437 20060101ALI20180306BHEP

Ipc: A61K 31/55 20060101ALI20180306BHEP

Ipc: A61K 31/44 20060101ALI20180306BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20190204

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 1153454

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190715

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602015033729

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20190710

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1153454

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191010

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191111

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191010

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191011

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20191110

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200224

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602015033729

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG2D Information on lapse in contracting state deleted

Ref country code: IS

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191031

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191031

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191021

26N No opposition filed

Effective date: 20200603

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20191031

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191031

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191021

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20151021

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190710

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602015033729

Country of ref document: DE

Owner name: MERCK SHARP & DOHME LLC, RAHWAY, US

Free format text: FORMER OWNER: MERCK SHARP & DOHME CORP., RAHWAY, N.J., US

REG Reference to a national code

Ref country code: GB

Ref legal event code: 732E

Free format text: REGISTERED BETWEEN 20221013 AND 20221019

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230914

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230914

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20230915

Year of fee payment: 9